You are on page 1of 3

EDITORIALS

Stem cell factor: the bridge between bone marrow adipocytes and hematopoietic cells
Ziru Li and Ormond A. MacDougald
Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
E-mail: ZIRU LI - liziru@umich.edu
doi:10.3324/haematol.2019.224188

W
hite adipocytes serve as an energy reservoir to in the bone marrow supernatant, which indicates that
store excessive calories in the form of lipid BMAT is a primary source of SCF in bone marrow.5
droplets and protect other tissues or organs from Deficiency of SCF in BMAT reduces the bone marrow cellu-
ectopic lipid accumulation. Brown adipocytes express larity, hematopoietic stem and progenitor cells (HSPC), com-
uncoupling protein 1 and are integral to adaptive thermoge- mon myeloid progenitors (CMP), megakaryocyte-erythro-
nesis. Whereas the functions of adipocytes in either white or cyte progenitor (MEP) and granulocyte-monocyte progeni-
brown adipose tissues are well documented, our knowledge tors (GMP) under steady-state condition. Consistent with
of bone marrow adipocytes (BMA) remains in its infancy. these changes in the progenitor cells of bone marrow, mice
Bone marrow adipose tissue (BMAT) occupies approximate- deficient for adipocyte SCF develop macrocytic anemia and
ly 50-70% of the bone marrow volume in human adults.1 It reduction of neutrophils, monocytes and lymphocytes in cir-
is a dynamic tissue and responds to multiple metabolic con- culation. In contrast to results in this study, Zhou et al.
ditions. For example, BMAT increases with obesity, aging, reported that the conditional deficiency of SCF in adipocytes
diabetes, caloric restriction, and irradiation.2 Although the driven by adiponectin-Cre/ER had no effect on
significance of BMAT expansion under these conditions is hematopoiesis under basal conditions.3 Although further
still largely unknown, BMA interact locally with hematopoi- investigation is necessary, the discrepancy between these
etic and bone cells, and contribute to global metabolism two studies might be due to the time-frame of SCF deletion,
through secretion of adiponectin, leptin, stem cell factor tamoxifen injection and/or animal lines. Of note, the dele-
(SCF), and other functional factors. For example, A-ZIP/F1 tion of SCF has no effect on the proliferation of HSPC evi-
mice, which lack adipose tissues throughout the body, denced by colony-forming assays, which suggests that
including BMAT, have delayed hematopoietic regeneration defects in BMAT-derived SCF influences the bone marrow
in long bones after irradiation.3 Our latest work also microenvironment rather than the intrinsic function of
observed that depletion of BMA by bariatric surgery is asso- HSPC.
ciated with a decrease in bone marrow erythroid cells and Since adiponectin-Cre is expressed in both peripheral
anemia.4 The importance of BMA and the derived factors on adipocytes and BMA, it is possible that there might be
hematopoiesis is further enhanced by a study in this issue of effects on hematopoiesis that are independent of BMAT. To
the Journal, in which Zhang et al.5 demonstrate that BMAT- more specifically study effects of BMA on the bone marrow
derived SCF mediates metabolic regulation of niche and hematopoiesis, Zhang et al. also deleted the Kitl
hematopoiesis. using osterix promoter, which traces BMA but not the other
Stem cell factor, also known as Kit ligand (Kitl), is a adipocytes. Again, knockout of Kitl from the osterix-positive
hematopoietic cytokine expressed in fibroblasts and (+) cells reduced bone marrow cellularity, hematopoietic pro-
endothelial cells, as well as in BMA.3 Together with its recep- genitor populations and mature blood cells including red
tor, c-Kit, SCF plays important roles in the maintenance of blood cells (RBC), neutrophils and monocytes, which is con-
hematopoietic stem cells (HSC) and hematopoiesis. sistent with the phenotypes from mice lacking adipocytic
Blockade of the interaction between c-Kit and SCF with anti- Kitl. Of note, in addition to BMA, osterix+ progenitors also
c-Kit antibody promotes the clearance of HSC, which indi- trace to osteoblasts.9,10 Mesenchymal and osteoblast lineage
cates the importance of Kitl/c-Kit signaling in HSC self- cells are involved in the maintenance and regulation of the
renewal.6 Loss-of-function mutations in c-Kit cause macro- supportive microenvironments necessary for quiescence,
cytic anemia, or even embryonic lethality under some severe self-renewal and differentiation of HSC.11,12 However, the
mutations.7 Inversely, mice with c-Kit gain-of-function SCF from osteoblasts is not required for HSC maintenance in
mutations developed erythrocytosis compatible with myelo- adult bone marrow under steady-state conditions.13
proliferative disorders.8 Analyses of multiple cell populations Although the possible effects of SCF derived from osterix+
isolated from bone marrow and adipose tissue have demon- progenitors on hematopoiesis could not be excluded and the
strated that BMA and LepR-positive (+) stromal cells are the bone phenotypes were not explored in this mouse model, it
primary sources of SCF, which is required for the regenera- should be appreciated that authors used both adiponectin-
tion of HSC and hematopoiesis after irradiation.3 Zhang et al. and osterix-driven Cre enzyme to confirm the phenotypes of
report that BMA-derived SCF is important for hematopoietic SCF-deficiency on hematopoiesis. These results strongly
homeostasis under basal (Figure 1), obese and aging condi- point to BMA as an important source of SCF since the com-
tions, and in response to β3-adrenergic agonists.5 mon cell type traced by adiponectin and osterix drivers is the
Knockout of SCF in adipocytes with an adiponectin driver BMA; however, development of BMA-specific transgenic
does not influence circulating SCF concentrations or pheno- mouse tools will be required to truly confirm these observa-
types of the peripheral adipose depots, which is perhaps due tions of BMA and the roles of SCF in the bone marrow niche
to compensatory expression of SCF from other sources, such homeostasis and hematopoiesis.
as endothelial cells, fibroblasts and stromal cells. The authors also investigated whether BMA-derived SCF
Interestingly, Zhang et al. observed a significant loss of SCF is required for hematopoietic adaptation to aging or high fat

haematologica | 2019; 104(9) 1689


Editorials

Figure 1. Bone marrow adipocytes influence the maintenance of hematopoietic stem cell (HSC) and hematopoiesis. Bone marrow cellularity is complex, but is main-
ly composed of hematopoietic cells and bone marrow adipocytes (BMA), which appear after birth and accumulate with age, obesity and irradiation. BMA originate
from osterix-positive (+) progenitor cells and secret adiponectin, stem cell factor (SCF) and other functional factors. In this study, Zhang et al.5 have demonstrated that
BMAT-derived SCF plays important roles in HSC maintenance and hematopoietic differentiation under baseline, aging and obese conditions. Deficiency of SCF in
BMAT hinders the self-renewal of HSC by influencing the bone marrow microenvironment and hematopoiesis through unknown mechanisms. RBC: red blood cell;
MPP: multipotent progenitor; CMP: common myeloid progenitor; MEP: megakaryocyte-erythrocyte progenitor; GMP: granulocyte-monocyte progenitor; CLP: common
lymphoid progenitor.

diet (HFD)-induced obesity. Whereas HFD, per se, did not models described above, it should be noted that alter-
increase the SCF concentrations in bone marrow super- ations of BMAT, SCF and hematopoiesis were not tightly
natant, this treatment increased bone marrow cellularity, associated under these conditions, which suggests that
HSPC, and mature blood cells, including granulocytes, hematopoietic metabolism is regulated by factors beyond
monocytes and lymphocytes, the effects of which were BMAT and its derived SCF. The global effects of obesity,
eliminated by SCF deficiency in adipocytes. Aging causes aging and β3-adrenoceptor activation cannot be excluded
similar increases in the HSPC, especially in the myeloid from this scenario. In addition, other secreted factors
lineage populations, and most of these effects required from BMAT may also play significant roles in
adipocyte-derived SCF. Further, these investigators hematopoiesis under these conditions. Unfortunately, the
explored a potential role for SCF in mediating effects of a secretome of BMAT remains largely unexplored.
β3-adrenergic receptor agonist. Activation of these recep- In summary, Zhang et al.5 have extended our under-
tors induces the lipolysis of white adipocytes, and while standing of the roles of BMAT in the bone marrow niche
although BMAT lipolysis is relatively resistant to β-adren- and the interaction between BMA and hematopoietic
ergic signaling,14 Zhang et al. observed that after adminis- cells. They thoroughly addressed their hypotheses using a
tration of a β3-adrenoceptor agonist, CL316, 243, SCF variety of animal models and complete profiling of
expression was increased in bone marrow without signif- hematopoietic changes. However, due to the complexity
icant changes in the BMA numbers.5 Consistent with the of whole-body metabolism and the lack of BMA-specific
elevated SCF in bone marrow, the numbers of HSPC, transgenic tools, further work will be required to deter-
including Lin-Sca1+c-Kit+ (LSK) cell, multipotent progeni- mine whether BMA-derived SCF regulates hematopoiesis
tor (MPP), MEP, GMP and CLP were increased by CL316, directly through Kitl/c-Kit signaling in hematopoietic cells
243 injection, the effects of which were compromised by or indirectly by changing the microenvironment of the
adipocyte-specific deficiency of SCF. Based on the animal bone marrow niche.

1690 haematologica | 2019; 104(9)


Editorials

Acknowledgments cell niches. Science. 2007;318(5854):1296-1299.


7. Nocka K, Majumder S, Chabot B, et al. Expression of c-kit gene
This work was supported by grants from the NIH to OAM products in known cellular targets of W mutations in normal and W
(R24 DK092759; R01 DK62876), and from the American mutant mice--evidence for an impaired c-kit kinase in mutant mice.
Diabetes Association to ZL (1-18-PDF-087). Genes Dev. 1989;3(6):816-826.
8. Bosbach B, Deshpande S, Rossi F, et al. Imatinib resistance and
microcytic erythrocytosis in a KitV558Delta;T669I/+ gatekeeper-
mutant mouse model of gastrointestinal stromal tumor. Proc Natl
References Acad Sci U S A. 2012;109(34):E2276-E2283.
9. Song L, Liu M, Ono N, Bringhurst FR, Kronenberg HM, Guo J. Loss
1. Cawthorn WP, Scheller EL, Learman BS, et al. Bone marrow adipose of wnt/beta-catenin signaling causes cell fate shift of preosteoblasts
tissue is an endocrine organ that contributes to increased circulating from osteoblasts to adipocytes. J Bone Miner Res. 2012;27(11):2344-
adiponectin during caloric restriction. Cell Metab. 2014;20(2):368- 2358.
375. 10. Mizoguchi T, Pinho S, Ahmed J, et al. Osterix marks distinct waves
2. Li Z, Hardij J, Bagchi DP, Scheller EL, MacDougald OA. of primitive and definitive stromal progenitors during bone marrow
Development, regulation, metabolism and function of bone marrow development. Dev Cell. 2014;29(3):340-349.
adipose tissues. Bone. 2018;110:134-140. 11. Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regu-
3. Zhou BO, Yu H, Yue R, et al. Bone marrow adipocytes promote the late the haematopoietic stem cell niche. Nature. 2003;425(6960):841-
regeneration of stem cells and haematopoiesis by secreting SCF. Nat 846.
Cell Biol. 2017;19(8):891-903. 12. Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL.
4. Li Z, Hardij J, Evers SS, et al. G-CSF partially mediates effects of Hematopoiesis is severely altered in mice with an induced osteoblast
sleeve gastrectomy on the bone marrow niche. J Clin Invest. deficiency. Blood. 2004;103(9):3258-3264.
2019;130:2404-2416. 13. Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and
5. Zhang Z, Huang Z, Ong B, Sahu C, Zeng H, Ruan HB. Bone marrow perivascular cells maintain haematopoietic stem cells. Nature.
adipose tissue-derived stem cell factor mediates metabolic regulation 2012;481(7382):457-462.
of hematopoiesis. Haematologica. 2019;104(9):1731-1743. 14. Scheller EL, Khandaker S, Learman BS, et al. Bone marrow
6. Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Efficient adipocytes resist lipolysis and remodeling in response to beta-adren-
transplantation via antibody-based clearance of hematopoietic stem ergic stimulation. Bone. 2019;118:32-41.

New potential players in hepcidin regulation


Maxwell Chappell and Stefano Rivella
Division of Hematology, Department of Pediatrics, Children’s Hospital of Philadelphia, Cell and Molecular Biology Graduate Group,
University of Pennsylvania, Abramson Research Center, Philadelphia, PA, USA
E-mail: STEFANO RIVELLA - rivellas@email.chop.edu
doi:10.3324/haematol.2019.224311

T
he manuscript by Liu and colleagues, published in Molecules such as HFE, transferrin receptor-2, and others
this issue of Haematologica, reports the identifica- communicate intracellularly when the transferrin satura-
tion of novel compounds able to increase hepcidin tion levels increase.3 It has been hypothesized that this
expression in normal mice as well as in animals affected sensing complex potentiates the SMAD complex activat-
by hemochromatosis and β-thalassemia intermedia (or ed by BMP6.5 Alternatively, or in addition, it has been
non-transfusion-dependent thalassemia) (Figure 1A).1 suggested that this complex acts upon hepcidin expres-
Hepcidin is the master regulator of iron secreted from sion by decreasing the ERK1/2 pathway.10
the liver and acts on ferroportin, a transmembrane pro- Under conditions that require enhanced red cell pro-
tein that functions as an iron exporter.2,3 Once hepcidin duction (as a consequence of a transient or chronic ane-
binds ferroportin, the complex is rapidly degraded, pre- mia), hepcidin synthesis is normally suppressed.2 A few
venting iron egress.2,3 Ferroportin is expressed in many factors have been identified that could play a role in this
types of cells, including enterocytes and macrophages.2,3 mechanism, such as erythroferrone and platelet-derived
Therefore, the relative abundance of hepcidin in the cir- growth factor BB.7,8 In particular, erythroferrone is secret-
culation and ferroportin on cell membranes control iron ed by erythroid cells and acts as a trap ligand, limiting the
absorption (from enterocytes) and iron recycling (from activity of BMP6 and other similar molecules.9
macrophages).2,3 Another player in the regulation of hepcidin is the mol-
Hepcidin expression is regulated by iron, inflammation ecule matriptase-2 (or TMPRSS6).2,3 This molecule pre-
and erythropoiesis.2,3 With regard to iron-mediated control vents hepcidin overexpression, which could lead to
of hepcidin, this is achieved through at least two mecha- hypoferremia and anemia.3,10 Although it is unclear which
nisms. The first senses the amount of intracellular iron in pathways and molecules control TMPRSS6, it has been
liver sinusoidal endothelial cells and responds by synthesiz- shown that TMPRSS6 is required for erythropoietin-
ing BMP6, and other similar ligands, belonging to the mediated hepcidin suppression in mice.11,12
TGFβ-like family.2-4 Increased intracellular concentration of In primary forms of hemochromatosis, patients show
iron leads to secretion of BMP6 from these cells.2-4 As a con- excessive iron absorption and suffer from iron overload
sequence, BMP6 binds and activates receptors that trigger (Figure 1A).7,8 This happens when hepcidin, or other
phosphorylation of a SMAD complex and stimulate hep- genes that control its expression, are mutated.2,3 In sec-
cidin expression in hepatic cells.2-4 ondary forms of hemochromatosis (as in β-thalassemia),
The second mechanism senses the iron in circulation the anemia triggers increased iron absorption, likely by
by recognizing iron-loaded transferrin molecules.3 increased expression of erythroferrone and other hypox-

haematologica | 2019; 104(9) 1691

You might also like